Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2009 Feb 24;46:S2–S14. doi: 10.1053/j.seminhematol.2008.12.005

Pathobiology of Secondary Immune Thrombocytopenia

Douglas B Cines a,, Howard Liebman b, Roberto Stasi c
PMCID: PMC2682438  NIHMSID: NIHMS99872  PMID: 19245930

Abstract

Primary immune thrombocytopenic purpura (ITP) remains a diagnosis of exclusion both from nonimmune causes of thrombocytopenia and immune thrombocytopenia that develops in the context of other disorders (secondary immune thrombocytopenia). The pathobiology, natural history, and response to therapy of the diverse causes of secondary ITP differ from each other and from primary ITP, so accurate diagnosis is essential. Immune thrombocytopenia can be secondary to medications or to a concurrent disease, such as an autoimmune condition (eg, systemic lupus erythematosus [SLE], antiphospholipid antibody syndrome [APS], immune thyroid disease, or Evans syndrome), a lymphoproliferative disease (eg, chronic lymphocytic leukemia or large granular T-lymphocyte lymphocytic leukemia), or chronic infection, eg, with Helicobacter pylori, human immunodeficiency virus (HIV), or hepatitis C virus (HCV). Response to infection may generate antibodies that cross-react with platelet antigens (HIV, H pylori) or immune complexes that bind to platelet Fcγ receptors (HCV), and platelet production may be impaired by infection of megakaryocyte (MK) bone marrow–dependent progenitor cells (HCV and HIV), decreased production of thrombopoietin (TPO), and splenic sequestration of platelets secondary to portal hypertension (HCV). Sudden and severe onset of thrombocytopenia has been observed in children after vaccination for measles, mumps, and rubella or natural viral infections, including Epstein-Barr virus, cytomegalovirus, and varicella zoster virus. This thrombocytopenia may be caused by cross-reacting antibodies and closely mimics acute ITP of childhood. Proper diagnosis and treatment of the underlying disorder, where necessary, play an important role in patient management.


Primary immune thrombocytopenic purpura (ITP) is a disorder of unknown etiology caused by antibody-, and possibly T-cell–mediated, platelet destruction by tissue macrophages and suppression of platelet production.1 ITP remains a diagnosis of exclusion.2, 3 This means not only excluding nonimmune causes of thrombocytopenia, but also other conditions in which an ITP-like syndrome is present, including those associated with autoimmune and lymphoproliferative disorders, acute and chronic infection, and certain drugs. Although these secondary forms of immune thrombocytopenia share with ITP the generation of self-reactive antiplatelet antibodies, they differ in specific aspects of pathobiology, natural history, and responsiveness to ITP-directed therapy. Moreover, optimal treatment strategies include management of the underlying disorder.

Normal Platelet Production

Platelet production starts with the complex bone marrow hematopoietic differentiation pathway known as megakaryopoieis.4 Pluripotent hematopoietic stem cells commit to differentiation as either a common lymphoid progenitor or common myeloid progenitor (CMP). The CMP then commits to another level of differentiation into either the granulocyte-macrophage progenitor or the megakaryocyte-erythroid progenitor (MEP). The signals that cue the stages of commitment are as yet only beginning to be understood. GATA-1, a 50-kd zinc finger DNA-binding protein encoded by an X-linked gene, acts in concert with another protein, friend of GATA (FOG), which affects transcription without binding to DNA.5, 6 Cells committed to the megakaryocytic lineage and thrombopoiesis, rather than erythropoiesis, begin to express CD41 and CD61 (integrin αIIbβ3 or αIIbIIIa), CD42 (glycoprotein [GP]Ib), and GPV.7, 8 Once committed, each primitive megakaryocyte (MK) progenitor or burst-forming unit (BFU-MK), a large complex with satellite collections of MKs, is capable, under the influence of thrombopoietin (TPO) and interleukin (IL)-3 and Steel factor, of producing up to several hundred mature MKs. Another form of mature progenitor, the colony-forming unit (CFU-MK) produces colonies that consist of 3 to 50 MKs responsive to TPO, although approximately 25% require both TPO and IL-3 to generate platelets.9, 10 Each MK produces up to about 4,000 platelets before undergoing apoptosis. A normal adult human produces about 1011 platelets daily, and this rate can be increased 20-fold with exogenous TPO.4

TPO and its receptor, c-Mpl, are the major identified regulators of MK and platelet production and also have key roles in the differentiation of hematopoietic stem cells. TPO signals via c-Mpl through the Jak-STAT,11, 12, 13, 14 Ras-Raf-MAPK,15 and PI3K pathways,16 which promote survival, proliferation, and polyploidy in MKs. In vitro TPO induces expression of c-MYC, a proto-oncogene active in various physiologic processes and in the dysregulation of MK production.17 The implications of this finding have yet to be addressed in vivo. The stages of thrombopoiesis include regulation of transcription, MK development, endomitotic spindling of the MK nucleus, cytoplasmic maturation, formation of active projections (protoplatelets) from the MK cytoplasm, extrusion of these protoplatelets from the bone marrow stroma into the circulation, and, lastly, separation and maturation into individual platelets.18

Primary ITP

Increased Platelet Destruction

There is extensive evidence that patients with ITP develop autoantibodies, generally IgG, that bind to platelets, which leads to their phagocytosis via Fcγ receptors expressed on tissue macrophages located predominantly in the spleen and liver.1, 19, 20, 21, 22 What provokes autoantibody production is unknown, but most ITP patients have antibodies against integrin αIIbβ3 (GPIIa/IIIb), GPIb/IX, or multiple platelet proteins by the time clinical disease, characterized by thrombocytopenia and mucocutaneous bleeding, is evident.23 Platelet destruction within macrophages or dendritic cells degrades platelet antigens to peptides. Peptides are expressed on the cell surface in the context of MHCII and costimulatory help for presentation to T cells, amplifying the initial immune response and possibly generating cryptic epitopes from other platelet glycoproteins, which spreads the immune response to involve multiple platelet antigens.24 ITP is characterized by reducing T-regulatory cells (reviewed in Stasi et al25) and Thy-2 cytokines,26 leading to a Thy1/Thy0 profile (reviewed in Ho-Yen et al27) and upregulation of costimulatory molecules28, 29 that facilitates proliferation of antigen-derived CD4-positive T cells and T-cell/B-cell cooperation to generate isotype-switched, high-affinity antibodies.30 There is emerging evidence that cytotoxic T cells are increased in the bone marrow31 and may contribute to platelet destruction32, 33 or impaired production (see below). The importance of platelet destruction in the periphery is affirmed by the fact that two thirds of patients develop and maintain remission after splenectomy, which curtails phagocytosis, but may also reduce antibody production over time. Likewise, most first- and second-line medical therapies for ITP are believed to work by impeding platelet destruction.2, 3

Decreased Platelet Production

For many years, it was assumed that platelet production increased dramatically in patients with ITP as a compensatory response to thrombocytopenia mediated by peripheral destruction. However, it has become apparent, based on studies of in vivo kinetics, that platelet production varies from mildly increased to mildly impaired in most patients with ITP.34, 35, 36 Synthesis of TPO in the liver is not regulated at the level of transcription.37 Plasma TPO levels in patients with ITP are normal to minimally increased38 as a result of increased clearance of the hormone bound to antibody-coated platelets and binding to an expanded MK mass.39 ITP antibodies, and possibly T cells,24 inhibit MK development in vitro 35, 36, 40 and may cause apoptosis and intramedullary destruction of platelets in vivo,41 contributing to failure of splenectomy and other treatments that act by inhibiting clearance. These findings also provide additional rationale for the effectiveness of TPO-receptor agonists.

Mechanisms of Immune Thrombocytopenia in Secondary ITP

Autoimmune Disorders

Systemic Lupus Erythematosus

Antinuclear antibodies are common in patients with ITP, but few develop systemic lupus erythematosus (SLE). However, approximately 20% to 25% of patients with SLE develop moderate-severe thrombocytopenia, which can be readily managed if immune-mediated or can be a marker of severe systemic disease.42 The pathogenesis of thrombocytopenia is multifactorial and includes: (1) antiplatelet glycoprotein antibodies as found in ITP; (2) immune complexes of diverse composition; (3) antiphospholipid antibodies (APLA) (see below); (4) vasculitis; (5) thrombotic microangiopathy; (6) hemophagocytosis; (7) autoantibodies to the c-Mpl receptor43 and MK; and (8) bone marrow stromal alterations44 not characteristic of ITP.45 Therefore, a thorough clinical and laboratory assessment is often required before a diagnosis of secondary immune thrombocytopenia should be entertained. It is also important to limit the use of corticosteroids and cytotoxic agents in these often heavily treated patients. Splenectomy may provide only transient benefit and is reserved for patients in whom severe thrombocytopenia is the predominant reason for treatment.46

Antiphospholpid Syndrome

APLA (lupus inhibitors and those that bind anionic phospholipids, beta-2-glycoprotein I [β2GPI], and prothrombin, among other specificities) are found in 20% to 70% of patients with ITP, depending on the thoroughness of the search, but their significance is debated and few patients develop antiphospholipid syndrome (APS).47, 48, 49, 50 The presence of APLA per se does not influence the effectiveness of ITP therapy in terms of inducing a platelet response. However, some, but not all, studies suggest such patients may be at risk for thrombosis once they do respond.47, 48, 49, 50, 51 The diagnosis is complicated by the fact that approximately 25% of patients with APS45 develop mild-moderate thrombocytopenia. Severe thrombocytopenia in APS correlates more closely with the presence of antiplatelet glycoprotein antibodies than either APLA or clinical manifestations.52, 53, 54, 55, 56 However, platelets do express receptors for β2GPI,57, 58, 59 which in theory could lead to APLA-mediated platelet activation alone or in association with other agonists,60 and there are anecdotal reports of response to aspirin, perhaps by inhibiting coagulation-mediated platelet consumption.61, 62, 63

Thyroid Disease

Mild-moderate thrombocytopenia is found commonly in patients with hyperthyroidism. Platelet survival is reduced, but returns to normal with restoration of the euthyroid state. Similarly, mild thrombocytopenia responsive to hormone replacement also occurs in some patients with hypothyroidism, possibly due to impaired production.64, 65 However, patients with immune thyroid disease develop immune thrombocytopenia requiring ITP-directed therapy more commonly than can be attributed to chance.66 Moreover, antithyroid antibodies occur commonly in adults and children with ITP,67, 68 leading some to recommend testing thyroid function in nonresponsive patients and prior to splenectomy.

Evans Syndrome

Patients with Evans syndrome (ES) develop immune hemolytic anemia, immune thrombocytopenia, and occasionally immune neutropenia.69, 70 The remarkable coincidence of two or three seemingly unrelated hematopoietic cell antibodies has now been associated with an underlying complex immunodeficiency in some patients (see below). Hemolysis may precede or follow the onset of thrombocytopenia and is typically more refractory to intervention, and the two cytopenias are often dyssynchronous in their manifestations. The response rates to ITP-directed therapy, including splenectomy, are less than in primary disease. More than 50% of children and some adults with ES have a clinical and pathological presentation that overlaps with the autoimmune lymphoproliferative syndrome (ALPS). ALPS is characterized by the chronic accumulation of nonmalignant lymphocytes leading to lymphadenopathy and (hepato)splenomegaly, >1% CD3+, CD4, CD8 (double-negative) T cells, impaired Fas-receptor/ligand mediated apoptosis in vitro due to mutations in Fas (CD95/Apo-1), or less commonly Fas ligand (Fas-L), caspase-8 or -10. Immune thrombocytopenia develops in about 20% of patients71, 72, 73, 74, 75, 76 and may respond relatively poorly to ITP therapies, although recent experience with rituximab and mycophenylate have been encouraging. Immune thrombocytopenia and ES also occur in approximately 10% to 15% of patients with common variable immune deficiency (CVID) and hypogammaglobulinemia. The onset of immune thrombocytopenia is typically in the third decade, although onsets from childhood to old age have been reported and typically precede the diagnosis of CVID by several years. The diagnosis should be sought in any patient with recurrent infection, as immunosuppressive therapy poses some risk and replacement with immune globulin is indicated.

Lymphoproliferative Disorders

There is an increased incidence of immune thrombocytopenia in patients with chronic lymphocytic leukemia (CLL),77 CD8 T-lymphocyte large granular lymphocytic leukemia (LGL),78 and possibly Hodgkin's disease.79, 80, 81 In CLL, it may be difficult to distinguish immune thrombocytopenia from marrow infiltration and splenomegaly82 or in the setting of treatment with fludarabine.83 Severe thrombocytopenia, which occurs in about 1% of patients with LGL, has been associated with clonal suppression of megakaryopoiesis.84, 85

Infectious Agents

Human Immunodeficiency Virus

The association between immune thrombocytopenia and the acquired immunodeficiency syndrome and subsequently as a presenting feature of HIV infection has been recognized since the early to mid 1980s.86, 87, 88 Thrombocytopenia is characterized both by an immune component similar in presentation and response to ITP, most evident in the early stages of disease,89 and progressive ineffective hematopoiesis with a decrease in platelet production as a result of MK infection90, 91, 92, 93 or marrow infiltration94, 95 as the disease progresses. HIV binds the CD4 receptor and coreceptors expressed on MKs,96, 97 is internalized,98, 99 and replicates within the infected cells100 leading to dysplasia, blebbing of the surface membrane, and vacuolization of peripheral cytoplasm.100, 101 The immune component is mediated through molecular mimicry involving anti-HIV antibodies that cross-react with platelet-membrane glycoproteins,102, 103, 103, 104, 105, 106 immune complexes,87, 107, 108, 109 and anti-GPIIIa49-66 antibodies that induce platelet lysis, at least in vitro, through a peroxidase-mediated pathway.106 Secondary causes of thrombocytopenia during HIV infection are generally the result of underlying opportunistic infections, malignancy, medications (eg, chemotherapeutic agents, interferon, and antiviral agents), or, less frequently, thrombotic microangiopathy.

HIV should be excluded in at-risk patients who present with ITP. Patients who present with immune thrombocytopenia early in the course of HIV infection respond to medical therapy (corticosteroids, intravenous anti-D, and intravenous immunoglobulin [IVIG]) and splenectomy as well as patients with ITP without proliferation of HIV infection or untoward incidence of opportunistic infection. Thrombocytopenia in patients with more advanced disease generally responds to highly active antiretroviral therapy.

Hepatitis C Virus

In some parts of the world, hepatitis C virus (HCV) infection has been detected in up to 30% of patients presenting with immune thrombocytopenia, even in the absence of overt hepatitis.110, 111, 112 The diagnosis of immune thrombocytopenia is confounded in patients with advanced liver disease because of hypersplenism113, 114 and decreased production of TPO.115, 116, 117, 118, 119 Antiplatelet antibodies are so common as to lack diagnostic utility.120 Possible mechanisms leading to immune destruction include binding of HCV followed by anti-HCV antibody to the platelet membrane, circulating anti-viral immune complexes,121, 122, 123 cross-reacting antibodies,123a and direct infection of MKs124 with expression of HCV RNA in platelets.125 Bone marrow production may be suppressed by HCV126 or interferon antiviral treatment.127 Patients typically present with significant bleeding in the presence of moderate thrombocytopenia.110 Optimal management involves suppression of viral replication. Use of TPO-receptor agonist may raise platelet counts sufficiently to permit sustained treatment with interferon-based therapy in a high proportion of patients.128

Helicobacter pylori

The success of eradicating infection with Helicobacter pylori among patients presenting with otherwise typical ITP varies from less than 1% to 5% in the United States to over 60% in Italy and Japan, with intermediate values reported from other countries.56, 129, 130 Several hypotheses relating H pylori to immune thrombocytopenia and to explain this variation have been proposed, including (1) regional differences in the expression of CagA-related genes,131, 132, 133 to which antibodies that cross-react with ITP platelets are generated through the process of molecular mimicry134; (2) cross-reactivity between H pylori cytotoxin-A protein and platelet antigens135; (3) adsorption to platelets of Lewis antigens, which are induced by H pylori in a strain-specific manner, where they are targets for anti-Lewis antibodies in patients with appropriate genetic backgrounds;136 (4) platelet activation and clearance through an interaction with H pylori–bound von Willebrand factor via platelet GPIb137; (5) somatic mutation of antibacterial antibodies from which antigen-independent autoantibodies emerge138; (6) monocytes from H pylori–positive patients demonstrate low levels of the inhibitory Fcγ receptor IIB and enhanced platelet phagocytosis, both of which are reversed after successful eradication,139 and (7) genetic variation, eg, the frequency of HLA-DRB*11,*14, and HLA-DQB1*03 is higher in patients with immune thrombocytopenia positive for H pylori than in those who are H pylori–negative, and those expressing HLA-DQB1*03 have a higher probability of a platelet response to eradication therapy.140 Of interest, titers of autoantibodies fall 12 to 24 weeks after successful eradication, whereas responses often occur in 1 to 2 weeks, suggesting additional mechanisms are operative.

Many, but not all studies indicate that H pylori is found more commonly in patients with disease that is milder in severity and of more recent onset.141 H pylori should be sought in all patients who come from regions where there is a strong association, but no consensus has emerged in the United States as to whether all ITP patients should be tested and/or treated.

Vaccination and Other Infections

Transient but severe thrombocytopenia occurs with an incidence of 1 in 25,000-40,000 vaccinations for measles, mumps, and rubella,142, 143, 144 and less commonly after vaccination against pneumococcus, Haemiphilus influenzae B, varicella zoster virus (VZV), and hepatitis B.145, 146 More than 80% of patients recover within 2 months, typically within 2 to 3 weeks,27, 30, 147, 148, 149 with less than 10% evolving into chronic ITP150 responsive to ITP-directed therapy. Thrombocytopenia occurs occasionally after naturally occurring infection with cytomegalovirus, rubella, Epstein-Barr virus, VZV, the severe acute respiratory syndrome coronavirus, and many others.151, 152, 153, 154 Thrombocytopenia may be immune, due to infection of MKs or progenitors, or result from peripheral consumption, eg, purpura fulminans due to VZV.

Post-transfusion Purpura

Post-transfusion purpura (PTP) is a rare but dramatic cause of immune thrombocytopenia. The typical patient is a multiparous or multitransfused female who presents with hemorrhage and the sudden onset of profound thrombocytopenia a mean of 7 days after transfusion of a blood product containing platelet antigens. The disease occurs primarily among the approximately 1% of Caucasians who are homozygous for HPA1b allele on GPIIIa, although other specificities have been reported. Patients not only develop anti-HPA1a antibodies, but also self-reactive antibodies of undetermined specificity, presumably as a result of epitope spread. Remarkably, PTP is not seen in women with neonatal alloimmune thrombocytopenia due to anti-HPA1a antibodies. Patients are at a high risk of bleeding and the clinical course is often protracted without therapy. IVIG is the standard of care. The utility of preventing subsequent exposure to the inciting antigen using washed HPA1b blood products is logical but unproven.

Drug-Induced Thrombocytopenia

The first case of drug-induced thrombocytopenia (DITP) was identified with quinine 140 years ago.155 DITP can result from bone marrow toxicity or immune-mediated destruction of platelets. Several hundred therapeutic agents have been implicated,156 but few reports are compelling.157 The diagnosis of DITP is generally based on this clinical scenario: (1) therapy with a candidate drug precedes thrombocytopenia by sufficient time to develop antibody; (2) there is no such temporal relationship with another drug; (3) all other reasonable causes have been excluded; (4) recovery occurs upon the discontinuation of the drug; and (5) re-exposure to the drug, if attempted, leads to recurrent thrombocytopenia. However, these criteria are rarely met, as underlying clinical circumstances are often complex and introduction and withdrawal of multiple drugs within a short time frame is common. Moreover, testing for drug-dependent antibodies is fraught with difficulty (solubility, concentration, effect on platelet activation, in vivo metabolism, lack of control patients on drug but without thrombocytopenia, etc), and few patients require rechallenge.

DITP typically affects only a small percentage of exposed patients, with the exception of heparin-induced thrombocytopenia (see below), and no known genetic predispositions or environmental criteria have been identified. Diverse mechanisms have been postulated to cause DITP, including immune complexes (heparin); induction of autoantibody (gold salts); anti–drug-specific antibodies (abciximab); drugs that induce conformational changes in platelet antigens that are recognized by antibody (fiban drugs); drug-induced antibodies that bind to platelet membranes in the presence of soluble drug (quinine); and hapten-dependent antibodies (some beta-lactam antibiotics)158, 157 (Table 1). Immune or nonimmune marrow suppression or nonimmune destruction (typified when ristocetin was introduced as an antibiotic), or thrombotic microangiopathy (eg, ticlopidine and possibly clopidogrel) occur less commonly.

Table 1.

Mechanisms Underlying Drug-Induced Thrombocytopenia

Classification (drugs) Mechanism Incidence
Hapten-dependent antibody (penicillin, some cephalosporin antibiotics) Hapten links covalently to membrane protein and induces drug-specific immune response Very rare
Quinine-type drug (quinine, sulfonamide antibiotics, nonsteroidal anti-inflammatory drugs) Drug induces antibody that binds to membrane protein in presence of soluble drug 26 cases per 1 million users of quinine per week, probably fewer cases with other drugs
Fiban-type drug (tirofiban, eptifibatide) Drug reacts with glycoprotein αIIbβ3 to induce a conformational change recognized by antibody (not yet confirmed) 0.2%-0.5%
Drug-specific antibody (abciximab) Antibody recognizes murine component of chimeric Fab fragment specific for platelet β3 0.5%-1.0% after first exposure,10%-14% after second exposure
Autoantibody (gold salts, procainamide) Drug induces antibody that reacts with autologous platelets in absence of drug 1.0% with gold, very rare with procainamide and other drugs
Immune complex (heparins) Drug binds to platelet factor 4, producing immune complex for which antibody is specific; immune complex activates platelets through Fc receptors 1%-3% among patients treated with unfractionated heparin for 7 days, rare with low-molecular-weight heparin

Table adapted from Aster and Bougie with permission from the Massachusetts Medical Society.157

Not surprisingly, most drug-dependent antibodies appear to recognize antigens involving prevalent platelet glycoprotein complexes such as αIIbβ3 or Ib/V/IX.159, 160 It has been hypothesized that drugs become immunogenic when they bind to a larger molecule, such as a protein generating antibodies to the drug itself or a drug-protein complex or a drug-induced alteration of a binding protein to induce neoepitopes.161 These hypotheses have been based primarily on the kinetics of drug-dependent inhibition of antibody binding to platelets or other cells rather than isolation of drug-protein, drug-antibody, or drug-protein-antibody complexes. Bougie and colleagues have proposed a model to reconcile existing hypotheses using quinine-dependent antibodies as a model.162 They posit that the drugs enhance the affinity of preexisting antiplatelet glycoprotein antibodies by providing a bridge between the complement determining region on the antiplatelet antibody with a drug-binding epitope on the platelet membrane. Whether the drug binds first to the antibody or to the platelet membrane protein depends on its relative affinity.

Thrombocytopenia Induced by Platelet Inhibitors

Tirofiban and eptifibatide used to prevent restenosis after coronary angioplasty can cause severe thrombocytopenia within hours of the first or subsequent doses. These ligand-mimetic drugs (fibans) competitively inhibit fibrinogen from binding to the platelet αIIbβ3 integrin receptor.163 These drugs may act as mixed agonists/antagonists. It is presumed that binding induces novel epitopes on αIIbβ3 that are recognized by preexisting (possibly preactivated) or drug-induced antibodies.

Abciximab is a chimeric human Fab fragment linked to specificity-determining sequences from a murine antibody to αIIbβ3 integrin that blocks fibrinogen binding. Sudden and often profound thrombocytopenia163, 164, 165 develops within hours in about 0.5% to 1% of patients treated with abciximab for the first time, and in approximately 10% of those treated a second time. Delayed onsets after first exposure occur less commonly. Most healthy individuals have naturally occurring antibodies to the C-terminus on human Fab (the papain cleavage site), which is present in the chimeric antibody. In contrast, patients who develop severe thrombocytopenia within a few hours of starting an infusion166 have preexisting antibodies that recognize the region that imparts GPIIIa specificity, although an effect of the drug on the conformation of GPIIbIIIa cannot be excluded.165 Abciximab-coated platelets may be detected for 10 to 14 days after treatment in some individuals, presumably due to coating of MKs or shuttling between destroyed and circulating platelets.167 IVIG and platelet transfusions have been used successfully to treat patients with profound abciximab- and fiban-induced thrombocytopenia.

Thrombocytopenia From Drug-Induced Autoantibodies

Rarely, drugs stimulate the formation of autoantibodies that target platelets in the absence of the inciting drug.168 Autoantibodies with an affinity for platelet GPV have developed in a few patients with rheumatoid arthritis treated with gold salts.169 Drug-independent autoantibodies have also been reported in occasional patients treated with quinine, procainamide, sulfonamide antibiotics, and interferons alfa and beta.157, 170 Thrombocytopenia is often protracted and may require ITP-directed therapy.

Heparin-Induced Thrombocytopenia

Heparin-induced thrombocytopenia (HIT) develops in 1% to 3% of patients who receive unfractionated heparin (UFH) intravenously in therapeutic doses for a minimum of 5 days. The prevalence is lower in patients treated exclusively with low-molecular-weight heparin or when UFH is given as thromboprophylaxis. The incidence is highest in patients undergoing cardiopulmonary bypass and orthopedic surgery, which is associated with intense platelet activation, inflammation, and underlying vascular disease, and lowest in children, during pregnancy, and patients receiving heparin during dialysis. Approximately 25% of patients develop arterial and/or venous thrombi,171 a number that may exceed 50% if the disease is not recognized and managed promptly.172 In treatment-naive subjects, an unexplained fall in the platelet count of greater than 40% begins 5 to 10 days after exposure; thrombosis typically follows soon after but can occur concomitantly or on occasion precede the fall in platelet count.173 Thrombocytopenia may occur within hours after exposure to heparin in patients who had been treated within the preceding 100 days in whom circulating antibodies may persist.174 Rarely, thrombocytopenia may be discovered or develop 1 to 2 weeks after the last known exposure (delayed HIT).175

HIT antibodies recognize oligomeric complexes formed between platelet factor 4 (PF4) released from activated platelets and heparin or glycosaminoglycans expressed on endothelium, monocytes, and platelets.176, 177, 178 Heparin/PF4-IgG complexes bind to FcγRIIa (CD32) on platelets, targeting them for clearance, but also stimulating cell activation with release of additional PF4. Binding of these PF4 followed by antibody to glycosaminoglycans on endothelial cells179 and monocytes180, 181 stimulates the expression of tissue factor, accelerating coagulation and reinforcing platelet activation. Heparin-independent anti-PF4 antibodies have been postulated to account for delayed HIT.175

Diagnosis rests on clinical recognition of the temporal relationship between heparin exposure and the signature clinical manifestation. Enzyme-linked immunosorbent assay (ELISA)-based assays that detect IgG, IgA, and IgM antibodies have a high sensitivity and negative predictive value but a false positive rate that can exceed 50% in patients post-bypass surgery,182 with unacceptably high false positive rates in other settings as well. The performance characteristics may be improved using newer assays that measure only IgG antibodies and using higher diagnostic cut-off optical density measurements, with little loss of negative predictive value.183 Assays based on platelet activation are more specific but less sensitive and the results are not routinely available in real time. Once the diagnosis is strongly suspected (moderate–high pretest probability), exposure to all forms of heparin (including flushes, heparin-bonded catheters, etc) must be discontinued and anticoagulation begun with either a direct thrombin inhibitor, such as hirudin or argatroban or an anti-Xa agent such as danaparoid, where available.184 Therapy reduces new thromboembolic events by approximately two thirds, but mortality rates and amputations due to preexisting clots are not affected. Therapy is continued until thrombocytopenia resolves and is overlapped with coumadin at a therapeutic international normalized ratio (INR) for 3 to 5 days. Anticoagulation is generally continued for 3 months, but may be extended depending on the underlying reason for the initial use of heparin and the sequelae of HIT-induced thrombosis.

Conclusion

There is compelling reason why the diagnosis of ITP requires the exclusion of other causes of thrombocytopenia, including conditions classified as secondary immune thrombocytopenia.185 The pathobiology of the various causes of secondary immune thrombocytopenia is often more complex than ITP (eg, marrow suppression or intravascular consumption due to clotting or vascular damage), they differ in natural history and responsiveness to ITP-directed therapy (eg, ES), and optimal therapy requires treatment of the underlying condition, eg, chronic infection, lymphoproliferative disease (CLL, LGL), and autoimmune conditions (eg, SLE, APS). Treatment of H pylori, HIV, or HCV may suffice to increase the platelet count and avoid protracted and sometimes ineffective and toxic treatments required to manage ITP. Drug-induced thrombocytopenias require recognition and withdrawal of the inciting agent. Posttransfusion purpura requires immediate recognition and treatment with IVIG. Patients with APS may be at risk for thrombosis. Treatment strategies for these forms of immune thrombocytopenia are discussed in greater detail elsewhere in this issue.

Footnotes

STATEMENT OF CONFLICT OF INTEREST: The authors disclose the following: Douglas B. Cines, MD: Consultant: Amgen, GSK, Syntonix, Bayer, Genzyme, Rigel; Howard A. Liebman, MD: Consultant: Amgen, GSK, Bristol-Myers Squibb; Grant/Research: Amgen, GSK, Bristol-Myers Squibb; Roberto Stasi, MD: Honoraria: Amgen, GSK; Speakers’ Bureau: Amgen, GSK.

References

  • 1.Cines D.B., Blanchette V.S. Immune thrombocytopenic purpura. N Engl J Med. 2002;346:995–1008. doi: 10.1056/NEJMra010501. [DOI] [PubMed] [Google Scholar]
  • 2.George J.N., Woolf S.H., Raskob G.E., Wasser J.S., Aledort L.M., Ballem P.J. Idiopathic thrombocytopenic purpura: a practice guideline developed by explicit methods for the American Society of Hematology. Blood. 1996;88:3–40. [PubMed] [Google Scholar]
  • 3.British Committee for Standards in Haematology General Haematology Task Force Guidelines for the investigation and management of idiopathic thrombocytopenic purpura in adults, children and in pregnancy. Br J Haematol. 2003;120:574–596. doi: 10.1046/j.1365-2141.2003.04131.x. [DOI] [PubMed] [Google Scholar]
  • 4.Kaushansky K. Historical review: megakaryopoiesis and thrombopoiesis. Blood. 2008;111:981–986. doi: 10.1182/blood-2007-05-088500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Martin D.I., Zon L.I., Mutter G., Orkin S.H. Expression of an erythroid transcription factor in megakaryocytic and mast cell lineages. Nature. 1990;344:444–447. doi: 10.1038/344444a0. [DOI] [PubMed] [Google Scholar]
  • 6.Tsang A.P., Visvader J.E., Turner C.A., Fujiwara Y., Yu C., Weiss M.J. FOG, a multitype zinc finger protein, acts as a cofactor for transcription factor GATA-1 in erythroid and megakaryocytic differentiation. Cell. 1997;90:109–119. doi: 10.1016/s0092-8674(00)80318-9. [DOI] [PubMed] [Google Scholar]
  • 7.Roth G.J., Yagi M., Bastian L.S. The platelet glycoprotein Ib-V-IX system: regulation of gene expression. Stem Cells. 1996;14(Suppl 1):188–193. doi: 10.1002/stem.5530140724. [DOI] [PubMed] [Google Scholar]
  • 8.Hodohara K., Fujii N., Yamamoto N., Kaushansky K. Stromal cell-derived factor-1 (SDF-1) acts together with thrombopoietin to enhance the development of megakaryocytic progenitor cells (CFU-MK) Blood. 2000;95:769–775. [PubMed] [Google Scholar]
  • 9.Broudy V.C., Lin N.L., Kaushansky K. Thrombopoietin (c-mpl ligand) acts synergistically with erythropoietin, stem cell factor, and interleukin-11 to enhance murine megakaryocyte colony growth and increases megakaryocyte ploidy in vitro. Blood. 1995;85:1719–1726. [PubMed] [Google Scholar]
  • 10.Kaushansky K., Broudy V.C., Lin N., Jorgensen M.J., McCarty J., Fox N. Thrombopoietin, the Mp1 ligand, is essential for full megakaryocyte development. Proc Natl Acad Sci U S A. 1995;92:3234–3238. doi: 10.1073/pnas.92.8.3234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Gurney A.L., Wong S.C., Henzel W.J., de Sauvage F.J. Distinct regions of c-Mpl cytoplasmic domain are coupled to the JAK-STAT signal transduction pathway and Shc phosphorylation. Proc Natl Acad Sci U S A. 1995;92:5292–5296. doi: 10.1073/pnas.92.12.5292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bacon C.M., Tortolani P.J., Shimosaka A., Rees R.C., Longo D.L., O'Shea J.J. Thrombopoietin (TPO) induces tyrosine phosphorylation and activation of STAT5 and STAT3. FEBS Lett. 1995;370:63–68. doi: 10.1016/0014-5793(95)00796-c. [DOI] [PubMed] [Google Scholar]
  • 13.Tortolani P.J., Johnston J.A., Bacon C.M., McVicar D.W., Shimosaka A., Linnekin D. Thrombopoietin induces tyrosine phosphorylation and activation of the Janus kinase, JAK2. Blood. 1995;85:3444–3451. [PubMed] [Google Scholar]
  • 14.Drachman J.G., Sabath D.F., Fox N.E., Kaushansky K. Thrombopoietin signal transduction in purified murine megakaryocytes. Blood. 1997;89:483–492. [PubMed] [Google Scholar]
  • 15.Nagata Y., Todokoro K. Thrombopoietin induces activation of at least two distinct signaling pathways. FEBS Lett. 1995;377:497–501. doi: 10.1016/0014-5793(95)01386-5. [DOI] [PubMed] [Google Scholar]
  • 16.Nakao T., Geddis A.E., Fox N.E., Kaushansky K. PI3K/Akt/FOXO3a pathway contributes to thrombopoietin-induced proliferation of primary megakaryocytes in vitro and in vivo via modulation of p27(Kip1) Cell Cycle. 2008;7:257–266. doi: 10.4161/cc.7.2.5148. [DOI] [PubMed] [Google Scholar]
  • 17.Chanprasert S., Geddis A.E., Barroga C., Fox N.E., Kaushansky K. Thrombopoietin (TPO) induces c-myc expression through a. Cell Signal. 2006;18:1212–1218. doi: 10.1016/j.cellsig.2005.09.010. [DOI] [PubMed] [Google Scholar]
  • 18.Battinelli E.M., Hartwig J.H., Italiano J.E., Jr Delivering new insight into the biology of megakaryopoiesis and thrombopoiesis. Curr Opin Hematol. 2007;14:419–426. doi: 10.1097/MOH.0b013e3282bad151. [DOI] [PubMed] [Google Scholar]
  • 19.Cines D.B., McMillan R. Pathogenesis of chronic immune thrombocytopenic purpura. Curr Opin Hematol. 2007;14:511–514. doi: 10.1097/MOH.0b013e3282ba5552. [DOI] [PubMed] [Google Scholar]
  • 20.Tavassoli M., McMillan R. Structure of the spleen in idiopathic thrombocytopenic purpura. Am J Clin Pathol. 1975;64:180–191. doi: 10.1093/ajcp/64.2.180. [DOI] [PubMed] [Google Scholar]
  • 21.Handin R.I., Stossel T.P. Phagocytosis of antibody-coated platelets by human granulocytes. N Engl J Med. 1974;290:989–993. doi: 10.1056/NEJM197405022901803. [DOI] [PubMed] [Google Scholar]
  • 22.McMillan R., Longmire R.L., Tavassoli M., Armstrong S., Yelenosky R. In vitro platelet phagocytosis by splenic leukocytes in idiopathic thrombocytopenic purpura. N Engl J Med. 1974;290:249–251. doi: 10.1056/NEJM197401312900505. [DOI] [PubMed] [Google Scholar]
  • 23.McMillan R. Autoantibodies and autoantigens in chronic immune thrombocytopenic purpura. Semin Hematol. 2000;37:239–248. doi: 10.1016/s0037-1963(00)90102-1. [DOI] [PubMed] [Google Scholar]
  • 24.He R., Reid D.M., Jones C.E., Shulman N.R. Spectrum of Ig classes, specificities, and titers of serum antiglycoproteins in chronic idiopathic thrombocytopenic purpura. Blood. 1994;83:1024–1032. [PubMed] [Google Scholar]
  • 25.Stasi R., Cooper N., Del Poeta G., Stipa E., Laura E.M., Abruzzese E. Analysis of regulatory T-cell changes in patients with idiopathic thrombocytopenic purpura receiving B cell-depleting therapy with rituximab. Blood. 2008;112:1147–1150. doi: 10.1182/blood-2007-12-129262. [DOI] [PubMed] [Google Scholar]
  • 26.Stasi R., Del Poeta G., Stipa E., Evangelista M.L., Trawinska M.M., Cooper N. Response to B-cell depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura. Blood. 2007;110:2924–2930. doi: 10.1182/blood-2007-02-068999. [DOI] [PubMed] [Google Scholar]
  • 27.Ho-Yen D.O., Hardie R., Sommerville R.G. Varicella-induced thrombocytopenia. J Infect. 1984;8:274–276. doi: 10.1016/s0163-4453(84)94147-1. [DOI] [PubMed] [Google Scholar]
  • 28.Solanilla A., Pasquet J.M., Viallard J.F., Contin C., Grosset C., Dechanet-Merville J. Platelet-associated CD154 in immune thrombocytopenic purpura. Blood. 2005;105:215–218. doi: 10.1182/blood-2003-07-2367. [DOI] [PubMed] [Google Scholar]
  • 29.Nagahama M., Nomura S., Kanazawa S., Ozaki Y., Kagawa H., Fukuhara S. Significance of chemokines and soluble CD40 ligand in patients with autoimmune thrombocytopenic purpura. Eur J Haematol. 2002;69:303–308. doi: 10.1034/j.1600-0609.2002.02774.x. [DOI] [PubMed] [Google Scholar]
  • 30.Roark J.H., Bussel J.B., Cines D.B., Siegel D.L. Genetic analysis of autoantibodies in idiopathic thrombocytopenic purpura reveals evidence of clonal expansion and somatic mutation. Blood. 2002;100:1388–1398. [PubMed] [Google Scholar]
  • 31.Olsson B., Ridell B., Carlsson L., Jacobsson S., Wadenvik H. Recruitment of T cells into bone marrow of ITP patients possibly due to elevated expression of VLA-4 and CX3CR1. Blood. 2008;112:1078–1084. doi: 10.1182/blood-2008-02-139402. [DOI] [PubMed] [Google Scholar]
  • 32.Olsson B., Andersson P.O., Jernas M., Jacobsson S., Carlsson B., Carlsson L.M. T-cell-mediated cytotoxicity toward platelets in chronic idiopathic thrombocytopenic purpura. Nat Med. 2003;9:1123–1124. doi: 10.1038/nm921. [DOI] [PubMed] [Google Scholar]
  • 33.Zhang F., Chu X., Wang L., Zhu Y., Li L., Ma D. Cell-mediated lysis of autologous platelets in chronic idiopathic thrombocytopenic purpura. Eur J Haematol. 2006;76:427–431. doi: 10.1111/j.1600-0609.2005.00622.x. [DOI] [PubMed] [Google Scholar]
  • 34.Stoll D., Cines D.B., Aster R.H., Murphy S. Platelet kinetics in patients with idiopathic thrombocytopenic purpura and moderate thrombocytopenia. Blood. 1985;65:584–588. [PubMed] [Google Scholar]
  • 35.McMillan R., Wang L., Tomer A., Nichol J., Pistillo J. Suppression of in vitro megakaryocyte production by antiplatelet autoantibodies from adult patients with chronic ITP. Blood. 2004;103:1364–1369. doi: 10.1182/blood-2003-08-2672. [DOI] [PubMed] [Google Scholar]
  • 36.Ballem P.J., Segal G.M., Stratton J.R., Gernsheimer T., Adamson J.W., Slichter S.J. Mechanisms of thrombocytopenia in chronic autoimmune thrombocytopenic purpura: Evidence of both impaired platelet production and increased platelet clearance. J Clin Invest. 1987;80:33–40. doi: 10.1172/JCI113060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Nagata Y., Shozaki Y., Nagahisa H., Nagasawa T., Abe T., Todokoro K. Serum thrombopoietin level is not regulated by transcription but by the total counts of both megakaryocytes and platelets during thrombocytopenia and thrombocytosis. Thromb Haemost. 1997;77:808–814. [PubMed] [Google Scholar]
  • 38.Kosugi S., Kurata Y., Tomiyama Y., Tahara T., Kato T., Tadokoro S. Circulating thrombopoietin level in chronic immune thrombocytopenic purpura. Br J Haematol. 1996;93:704–706. doi: 10.1046/j.1365-2141.1996.d01-1702.x. [DOI] [PubMed] [Google Scholar]
  • 39.Emmons R.V., Reid D.M., Cohen R.L., Meng G., Young N.S., Dunbar C.E. Human thrombopoietin levels are high when thrombocytopenia is due to megakaryocyte deficiency and low when due to increased platelet destruction. Blood. 1996;87:4068–4071. [PubMed] [Google Scholar]
  • 40.Chang M., Nakagawa P.A., Williams S.A., Schwartz M.R., Imfeld K.L., Buzby J.S. Immune thrombocytopenic purpura (ITP) plasma and purified ITP monoclonal autoantibodies inhibit megakaryocytopoiesis in vitro. Blood. 2003;102:887–895. doi: 10.1182/blood-2002-05-1475. [DOI] [PubMed] [Google Scholar]
  • 41.Houwerzijl E.J., Blom N.R., van der Want J.J., Esselink M.T., Koornstra J.J., Smit J.W. Ultrastructural study shows morphologic features of apoptosis and para-apoptosis in megakaryocytes from patients with idiopathic thrombocytopenic purpura. Blood. 2004;103:500–506. doi: 10.1182/blood-2003-01-0275. [DOI] [PubMed] [Google Scholar]
  • 42.Nossent J.C., Swaak A.J. Prevalence and significance of haematological abnormalities in patients with systemic lupus erythematosus. Q J Med. 1991;80:605–612. [PubMed] [Google Scholar]
  • 43.Kuwana M., Okazaki Y., Kajihara M., Kaburaki J., Miyazaki H., Kawakami Y. Autoantibody to c-Mpl (thrombopoietin receptor) in systemic lupus erythematosus: relationship to thrombocytopenia with megakaryocytic hypoplasia. Arthritis Rheum. 2002;46:2148–2159. doi: 10.1002/art.10420. [DOI] [PubMed] [Google Scholar]
  • 44.Ziakas P.D., Routsias J.G., Giannouli S., Tasidou A., Tzioufas A.G., Voulgarelis M. Suspects in the tale of lupus-associated thrombocytopenia. Clin Exp Immunol. 2006;145:71–80. doi: 10.1111/j.1365-2249.2006.03122.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gernsheimer T. Epidemiology and pathophysiology of immune thrombocytopenic purpura. Eur J Haematol. 2008;(Suppl):3–8. doi: 10.1111/j.1600-0609.2007.00998.x. [DOI] [PubMed] [Google Scholar]
  • 46.Arnal C., Piette J.C., Leone J., Taillan B., Hachulla E., Roudot-Thoraval F. Treatment of severe immune thrombocytopenia associated with systemic lupus erythematosus: 59 cases. J Rheumatol. 2002;29:75–83. [PubMed] [Google Scholar]
  • 47.Stasi R., Stipa E., Masi M., Oliva F., Sciarra A., Perrotti A. Prevalence and clinical significance of elevated antiphospholipid antibodies in patients with idiopathic thrombocytopenic purpura. Blood. 1994;84:4203–4208. [PubMed] [Google Scholar]
  • 48.Diz-Kucukkaya R., Hacihanefioglu A., Yenerel M., Turgut M., Keskin H., Nalcaci M. Antiphospholipid antibodies and antiphospholipid syndrome in patients presenting with immune thrombocytopenic purpura: a prospective cohort study. Blood. 2001;98:1760–1764. doi: 10.1182/blood.v98.6.1760. [DOI] [PubMed] [Google Scholar]
  • 49.Dash S., Marwaha R.K., Mohanty S. Lupus anticoagulant in immune thrombocytopenic purpura. Indian J Pediatr. 2004;71:505–507. doi: 10.1007/BF02724290. [DOI] [PubMed] [Google Scholar]
  • 50.Bidot C.J., Jy W., Horstman L.L., Ahn E.R., Yaniz M., Ahn Y.S. Antiphospholipid antibodies (APLA) in immune thrombocytopenic purpura (ITP) and antiphospholipid syndrome (APS) Am J Hematol. 2006;81:391–396. doi: 10.1002/ajh.20571. [DOI] [PubMed] [Google Scholar]
  • 51.Bidot C.J., Jy W., Horstman L.L., Ahn E.R., Jimenez J.J., Yaniz M. Antiphospholipid antibodies in immune thrombocytopenic purpura tend to emerge in exacerbation and decline in remission. Br J Haematol. 2006;128:366–372. doi: 10.1111/j.1365-2141.2004.05314.x. [DOI] [PubMed] [Google Scholar]
  • 52.Galli M., Daldossi M., Barbui T. Anti-glycoprotein Ib/IX and IIb/IIIa antibodies in patients with antiphospholipid antibodies. Thromb Haemost. 1994;71:571–575. [PubMed] [Google Scholar]
  • 53.Macchi L., Rispal P., Clofent-Sanchez G., Pellegrin J.L., Nurden P., Leng B. Anti-platelet antibodies in patients with systemic lupus erythematosus and the primary antiphospholipid antibody syndrome: their relationship with the observed thrombocytopenia. Br J Haematol. 1997;98:336–341. doi: 10.1046/j.1365-2141.1997.2243038.x. [DOI] [PubMed] [Google Scholar]
  • 54.Godeau B., Piette J.C., Fromont P., Intrator L., Schaeffer A., Bierling P. Specific antiplatelet glycoprotein autoantibodies are associated with the thrombocytopenia of primary antiphospholipid syndrome. Br J Haematol. 1997;98:873–879. doi: 10.1046/j.1365-2141.1997.3063123.x. [DOI] [PubMed] [Google Scholar]
  • 55.Fabris F., Steffan A., Cordiano I., Borzini P., Luzzatto G., Randi M.L. Specific antiplatelet autoantibodies in patients with antiphospholipid antibodies and thrombocytopenia. Eur J Haematol. 1994;53:232–236. doi: 10.1111/j.1600-0609.1994.tb00195.x. [DOI] [PubMed] [Google Scholar]
  • 56.Liebman H.A., Stasi R. Secondary immune thrombocytopenic purpura. Curr Opin Hematol. 2007;14:557–573. doi: 10.1097/MOH.0b013e3282ab9904. [DOI] [PubMed] [Google Scholar]
  • 57.Pennings M.T., Derksen R.H., Urbanus R.T., Tekelenburg W.L., Hemrika W., De Groot P.G. Platelets express three different splice variants of ApoER2 that are all involved in signaling. J Thromb Haemost. 2007;5:1538–1544. doi: 10.1111/j.1538-7836.2007.02605.x. [DOI] [PubMed] [Google Scholar]
  • 58.Pennings M.T., Derksen R.H., van L.M., Adelmeijer J., Vanhoorelbeke K., Urbanus R.T. Platelet adhesion to dimeric beta-glycoprotein I under conditions of flow is mediated by at least two receptors: glycoprotein Ibalpha and apolipoprotein E receptor 2′. J Thromb Haemost. 2007;5:369–377. doi: 10.1111/j.1538-7836.2007.02310.x. [DOI] [PubMed] [Google Scholar]
  • 59.Shi T., Giannakopoulos B., Yan X., Yu P., Berndt M.C., Andrews R.K. Anti-beta2-glycoprotein I antibodies in complex with beta2-glycoprotein I can activate platelets in a dysregulated manner via glycoprotein Ib-IX-V. Arthritis Rheum. 2006;54:2558–2567. doi: 10.1002/art.21968. [DOI] [PubMed] [Google Scholar]
  • 60.Nojima J., Suehisa E., Kuratsune H., Machii T., Koike T., Kitani T. Platelet activation induced by combined effects of anticardiolipin and lupus anticoagulant IgG antibodies in patients with systemic lupus erythematosus—possible association with thrombotic and thrombocytopenic complications. Thromb Haemost. 1999;81:436–441. [PubMed] [Google Scholar]
  • 61.Alliot C., Messouak D., Albert F., Barrios M. Correction of thrombocytopenia with aspirin in the primary antiphospholipid syndrome. Am J Hematol. 2001;68:215. doi: 10.1002/ajh.1182. [DOI] [PubMed] [Google Scholar]
  • 62.Alarcon-Segovia D., Sanchez-Guerrero J. Correction of thrombocytopenia with small dose aspirin in the primary antiphospholipid syndrome. J Rheumatol. 1989;16:1359–1361. [PubMed] [Google Scholar]
  • 63.Cohen M.G., Lui S.F. Multiple complications of the antiphospholipid syndrome with apparent response to aspirin therapy. J Rheumatol. 1992;19:803–806. [PubMed] [Google Scholar]
  • 64.Panzer S., Haubenstock A., Minar E. Platelets in hyperthyroidism: studies on platelet counts, mean platelet volume, 111-indium-labeled platelet kinetics, and platelet-associated immunoglobulins G and M. J Clin Endocrinol Metab. 1990;70:491–496. doi: 10.1210/jcem-70-2-491. [DOI] [PubMed] [Google Scholar]
  • 65.Stiegler G., Stohlawetz P., Brugger S., Jilma B., Vierhapper H., Hocker P. Elevated numbers of reticulated platelets in hyperthyroidism: direct evidence for an increase of thrombopoiesis. Br J Haematol. 1998;101:656–658. doi: 10.1046/j.1365-2141.1998.00765.x. [DOI] [PubMed] [Google Scholar]
  • 66.Cordiano I., Betterle C., Spadaccino C.A., Soini B., Girolami A., Fabris F. Autoimmune thrombocytopenia (AITP) and thyroid autoimmune disease (TAD): overlapping syndromes? Clin Exp Immunol. 1998;113:373–378. doi: 10.1046/j.1365-2249.1998.00677.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ioachimescu A.G., Makdissi A., Lichtin A., Zimmerman R.S. Thyroid disease in patients with idiopathic thrombocytopenia: a cohort study. Thyroid. 2007;17:1137–1142. doi: 10.1089/thy.2007.0066. [DOI] [PubMed] [Google Scholar]
  • 68.Pratt E.L., Tarantino M.D., Wagner D., Hirsch Pescovitz O., Bowyer S., Shapiro A.D. Prevalence of elevated antithyroid antibodies and antinuclear antibodies in children with immune thrombocytopenic purpura. Am J Hematol. 2005;79:175–179. doi: 10.1002/ajh.20299. [DOI] [PubMed] [Google Scholar]
  • 69.Pegels J.G., Helmerhorst F.M., van Leeuwen E.F., van de Plas-van Dalen C., Engelfriet C.P., von dem Borne A.E. The Evans syndrome: characterization of the responsible autoantibodies. Br J Haematol. 1982;51:445–450. doi: 10.1111/j.1365-2141.1982.tb02801.x. [DOI] [PubMed] [Google Scholar]
  • 70.Miller B.A., Shultz Beardsley D. Autoimmune pancytopenia of childhood associated with multisystem disease manifestations. J Pediatr. 1983;103:877–881. doi: 10.1016/s0022-3476(83)80705-7. [DOI] [PubMed] [Google Scholar]
  • 71.Wang W., Herrod H., Pui C.H., Presbury G., Wilimas J. Immunoregulatory abnormalities in Evans syndrome. Am J Hematol. 1983;15:381–390. doi: 10.1002/ajh.2830150409. [DOI] [PubMed] [Google Scholar]
  • 72.Fisher G.H., Rosenberg F.J., Straus S.E., Dale J.K., Middleton L.A., Lin A.Y. Dominant interfering Fas gene mutations impair apoptosis in a human autoimmune lymphoproliferative syndrome. Cell. 1995;81:935–946. doi: 10.1016/0092-8674(95)90013-6. [DOI] [PubMed] [Google Scholar]
  • 73.Rieux-Laucat F., Le Deist F., Hivroz C., Roberts I.A., Debatin K.M., Fischer A. Mutations in Fas associated with human lymphoproliferative syndrome and autoimmunity. Science. 1995;268:1347–1349. doi: 10.1126/science.7539157. [DOI] [PubMed] [Google Scholar]
  • 74.Le Deist F., Emile J.F., Rieux-Laucat F., Benkerrou M., Roberts I. Clinical, immunological, and pathological consequences of Fas-deficient conditions. Lancet. 1996;348:719–723. doi: 10.1016/S0140-6736(96)02293-3. [DOI] [PubMed] [Google Scholar]
  • 75.Rieux-Laucat F., Le Deist F., Fischer A. Autoimmune lymphoproliferative syndromes: genetic defects of apoptosis pathways. Cell Death Differ. 2003;10:124–133. doi: 10.1038/sj.cdd.4401190. [DOI] [PubMed] [Google Scholar]
  • 76.Teachey D.T., Manno C.S., Axsom K.M., Andrews T., Choi J.K., Greenbaum B.H. Unmasking Evans syndrome: T-cell phenotype and apoptotic response reveal autoimmune lymphoproliferative syndrome (ALPS) Blood. 2005;105:2443–2448. doi: 10.1182/blood-2004-09-3542. [DOI] [PubMed] [Google Scholar]
  • 77.Hamblin T.J., Oscier D.G., Young B.J. Autoimmunity in chronic lymphocytic leukaemia. J Clin Pathol. 1986;39:713–716. doi: 10.1136/jcp.39.7.713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Dhodapkar M.V., Li C.Y., Lust J.A., Tefferi A., Phyliky R.L. Clinical spectrum of clonal proliferations of T-large granular lymphocytes: a T-cell clonopathy of undetermined significance? Blood. 1994;84:1620–1627. [PubMed] [Google Scholar]
  • 79.Xiros N., Binder T., Anger B., Bohlke J., Heimpel H. Idiopathic thrombocytopenic purpura and autoimmune hemolytic anemia in Hodgkin's disease. Eur J Haematol. 1988;40:437–441. doi: 10.1111/j.1600-0609.1988.tb00853.x. [DOI] [PubMed] [Google Scholar]
  • 80.Landgren O., Engels E.A., Pfeiffer R.M., Gridley G., Mellemkjaer L., Olsen J.H. Autoimmunity and susceptibility to Hodgkin lymphoma: a population-based case-control study in Scandinavia. J Natl Cancer Inst. 2006;98:1321–1330. doi: 10.1093/jnci/djj361. [DOI] [PubMed] [Google Scholar]
  • 81.Martinelli G., Zinzani P.L., Magagnoli M., Vianelli N., Tura S. Incidence and prognostic significance of idiopathic thrombocytopenic purpura in patients with Hodgkin's disease in complete hematological remission. Haematologica. 1998;83:669–670. [PubMed] [Google Scholar]
  • 82.Hamblin T.J. Autoimmune complications of chronic lymphocytic leukemia. Semin Oncol. 2006;33:230–239. doi: 10.1053/j.seminoncol.2006.01.011. [DOI] [PubMed] [Google Scholar]
  • 83.Montillo M., Tedeschi A., Leoni P. Recurrence of autoimmune thrombocytopenia after treatment with fludarabine in a patient with chronic lymphocytic leukemia. Leuk Lymphoma. 1994;15:187–188. doi: 10.3109/10428199409051696. [DOI] [PubMed] [Google Scholar]
  • 84.Ergas D., Tsimanis A., Shtalrid M., Duskin C., Berrebi A. T-gamma large granular lymphocyte leukemia associated with amegakaryocytic thrombocytopenic purpura, Sjogren's syndrome, and polyglandular autoimmune syndrome type II, with subsequent development of pure red cell aplasia. Am J Hematol. 2002;69:132–134. doi: 10.1002/ajh.10024. [DOI] [PubMed] [Google Scholar]
  • 85.Lai D.W., Loughran T.P., Jr, Maciejewski J.P., Sasu S., Song S.X., Epling-Burnette P.K. Acquired amegakaryocytic thrombocytopenia and pure red cell aplasia associated with an occult large granular lymphocyte leukemia. Leuk Res. 2008;32:823–827. doi: 10.1016/j.leukres.2007.08.012. [DOI] [PubMed] [Google Scholar]
  • 86.Morris L., Distenfeld A., Amorosi E., Karpatkin S. Autoimmune thrombocytopenic purpura in homosexual men. Ann Intern Med. 1982;96:714–717. doi: 10.7326/0003-4819-96-6-714. [DOI] [PubMed] [Google Scholar]
  • 87.Walsh C.M., Nardi M.A., Karpatkin S. On the mechanism of thrombocytopenic purpura in sexually active homosexual men. N Engl J Med. 1984;311:635–639. doi: 10.1056/NEJM198409063111004. [DOI] [PubMed] [Google Scholar]
  • 88.Abrams D.I., Kiprov D.D., Goedert J.J., Sarngadharan M.G., Gallo R.C., Volberding P.A. Antibodies to human T-lymphotropic virus type III and development of the acquired immunodeficiency syndrome in homosexual men presenting with immune thrombocytopenia. Ann Intern Med. 1986;104:47–50. doi: 10.7326/0003-4819-104-1-47. [DOI] [PubMed] [Google Scholar]
  • 89.Murphy M.F., Metcalfe P., Waters A.H., Carne C.A., Weller I.V., Linch D.C. Incidence and mechanism of neutropenia and thrombocytopenia in patients with human immunodeficiency virus infection. Br J Haematol. 1987;66:337–340. doi: 10.1111/j.1365-2141.1987.tb06920.x. [DOI] [PubMed] [Google Scholar]
  • 90.Zon L.I., Arkin C., Groopman J.E. Haematologic manifestations of the human immune deficiency virus (HIV) Br J Haematol. 1987;66:251–256. doi: 10.1111/j.1365-2141.1987.tb01307.x. [DOI] [PubMed] [Google Scholar]
  • 91.Ballem P.J., Belzberg A., Devine D.V., Lyster D., Spruston B., Chambers H. Kinetic studies of the mechanism of thrombocytopenia in patients with human immunodeficiency virus infection. N Engl J Med. 1992;327:1779–1784. doi: 10.1056/NEJM199212173272503. [DOI] [PubMed] [Google Scholar]
  • 92.Landonio G., Nosari A., Spinelli F., Vigorelli R., Caggese L., Schlacht I. HIV-related thrombocytopenia: four different clinical subsets. Haematologica. 1992;77:398–401. [PubMed] [Google Scholar]
  • 93.Najean Y., Rain J.D. The mechanism of thrombocytopenia in patients with HIV infection. J Lab Clin Med. 1994;123:415–420. [PubMed] [Google Scholar]
  • 94.Bahner I., Kearns K., Coutinho S., Leonard E.H., Kohn D.B. Infection of human marrow stroma by human immunodeficiency virus-1 (HIV-1) is both required and sufficient for HIV-1-induced hematopoietic suppression in vitro: demonstration by gene modification of primary human stroma. Blood. 1997;90:1787–1798. [PubMed] [Google Scholar]
  • 95.Moses A, Nelson J, Bagby GC Jr. The influence of human immunodeficiency virus-1 on hematopoiesis. Blood. 198;91:1479-95. [PubMed]
  • 96.Kowalska M.A., Ratajczak J., Hoxie J., Brass L.F., Gewirtz A., Poncz M. Megakaryocyte precursors, megakaryocytes and platelets express the HIV co-receptor CXCR4 on their surface: determination of response to stromal-derived factor-1 by megakaryocytes and platelets. Br J Haematol. 1999;104:220–229. doi: 10.1046/j.1365-2141.1999.01169.x. [DOI] [PubMed] [Google Scholar]
  • 97.Sato T., Sekine H., Kakuda H., Miura N., Sunohara M., Fuse A. HIV infection of megakaryocytic cell lines. Leuk Lymphoma. 2000;36:397–404. doi: 10.3109/10428190009148861. [DOI] [PubMed] [Google Scholar]
  • 98.Zucker-Franklin D., Seremetis S., Zheng Z.Y. Internalization of human immunodeficiency virus type I and other retroviruses by megakaryocytes and platelets. Blood. 1990;75:1920–1923. [PubMed] [Google Scholar]
  • 99.Sakaguchi M., Sato T., Groopman J.E. Human immunodeficiency virus infection of megakaryocytic cells. Blood. 1991;77:481–485. [PubMed] [Google Scholar]
  • 100.Zucker-Franklin D., Cao Y.Z. Megakaryocytes of human immunodeficiency virus-infected individuals express viral RNA. Proc Natl Acad Sci U S A. 1989;86:5595–5599. doi: 10.1073/pnas.86.14.5595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zucker-Franklin D., Termin C.S., Cooper M.C. Structural changes in the megakaryocytes of patients infected with the human immune deficiency virus (HIV-1) Am J Pathol. 1989;134:1295–1303. [PMC free article] [PubMed] [Google Scholar]
  • 102.Karpatkin S., Nardi M.A., Hymes K.B. Sequestration of anti-platelet GPIIIa antibody in rheumatoid factor immune complexes of human immunodeficiency virus 1 thrombocytopenic patients. Proc Natl Acad Sci U S A. 1995;92:2263–2267. doi: 10.1073/pnas.92.6.2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li Z., Nardi M.A., Karpatkin S. Role of molecular mimicry to HIV-1 peptides in HIV-1-related immunologic thrombocytopenia. Blood. 2005;106:572–576. doi: 10.1182/blood-2005-01-0243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Bettaieb A., Fromont P., Louache F., Oksenhendler E., Vainchenker W., Duedari N. Presence of cross-reactive antibody between human immunodeficiency virus (HIV) and platelet glycoproteins in HIV-related immune thrombocytopenic purpura. Blood. 1992;80:162–169. [PubMed] [Google Scholar]
  • 105.Hohmann A.W., Booth K., Peters V., Gordon D.L., Comacchio R.M. Common epitope on HIV p24 and human platelets. Lancet. 1993;342:1274–1275. doi: 10.1016/0140-6736(93)92363-x. [DOI] [PubMed] [Google Scholar]
  • 106.Nardi M., Tomlinson S., Greco M.A., Karpatkin S. Complement-independent, peroxide-induced antibody lysis of platelets in HIV-1-related immune thrombocytopenia. Cell. 2001;106:551–561. doi: 10.1016/s0092-8674(01)00477-9. [DOI] [PubMed] [Google Scholar]
  • 107.Karpatkin S., Nardi M., Lennette E.T., Byrne B., Poiesz B. Anti-human immunodeficiency virus type 1 antibody complexes on platelets of seropositive thrombocytopenic homosexuals and narcotic addicts. Proc Natl Acad Sci U S A. 1988;85:9763–9767. doi: 10.1073/pnas.85.24.9763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Karpatkin S., Nardi M. Autoimmune anti-HIV-1gp120 antibody with antiidiotype-like activity in sera and immune complexes of HIV-1-related immunologic thrombocytopenia. J Clin Invest. 1992;89:356–364. doi: 10.1172/JCI115593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Koefoed K., Ditzel H.J. Identification of talin head domain as an immunodominant epitope of the antiplatelet antibody response in patients with HIV-1-associated thrombocytopenia. Blood. 2004;104:4054–4062. doi: 10.1182/blood-2004-01-0386. [DOI] [PubMed] [Google Scholar]
  • 110.Rajan S.K., Espina B.M., Liebman H.A. Hepatitis C virus-related thrombocytopenia: clinical and laboratory characteristics compared with chronic immune thrombocytopenic purpura. Br J Haematol. 2005;129:818–824. doi: 10.1111/j.1365-2141.2005.05542.x. [DOI] [PubMed] [Google Scholar]
  • 111.Pyrsopoulos N.T., Reddy K.R. Extrahepatic manifestations of chronic viral hepatitis. Curr Gastroenterol Rep. 2001;3:71–78. doi: 10.1007/s11894-001-0044-1. [DOI] [PubMed] [Google Scholar]
  • 112.Pockros P.J., Duchini A., McMillan R., Nyberg L.M., McHutchison J., Viernes E. Immune thrombocytopenic purpura in patients with chronic hepatitis C virus infection. Am J Gastroenterol. 2002;97:2040–2045. doi: 10.1111/j.1572-0241.2002.05845.x. [DOI] [PubMed] [Google Scholar]
  • 113.Yabu K., Kiyosawa K., Ako S., Usuda S., Yoshizawa K., Tanaka E. Type C chronic hepatitis associated with thrombocytopenia in two patients. J Gastroenterol Hepatol. 1994;9:99–104. doi: 10.1111/j.1440-1746.1994.tb01223.x. [DOI] [PubMed] [Google Scholar]
  • 114.Aster R.H. Pooling of platelets in the spleen: role in the pathogenesis of “hypersplenic” thrombocytopenia. J Clin Invest. 1966;45:645–657. doi: 10.1172/JCI105380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Espanol I., Gallego A., Enriquez J., Rabella N., Lerma E., Hernandez A. Thrombocytopenia associated with liver cirrhosis and hepatitis C viral infection: role of thrombopoietin. Hepatogastroenterology. 2000;47:1404–1406. [PubMed] [Google Scholar]
  • 116.Adinolfi L.E., Giordano M.G., Andreana A., Tripodi M.F., Utili R., Cesaro G. Hepatic fibrosis plays a central role in the pathogenesis of thrombocytopenia in patients with chronic viral hepatitis. Br J Haematol. 2001;113:590–595. doi: 10.1046/j.1365-2141.2001.02824.x. [DOI] [PubMed] [Google Scholar]
  • 117.Jiang X.H., Xie Y.T., Tan D.M. [Study on the influencing factors of thrombocytopenia in viral hepatitis] Zhonghua Gan Zang Bing Za Zhi. 2004;12:734–736. [PubMed] [Google Scholar]
  • 118.Peck-Radosavljevic M., Zacherl J., Meng Y.G. Is inadequate thrombopoietin production a major cause of thrombocytopenia in cirrhosis of the liver? J Hepatol. 1997;27:127–131. doi: 10.1016/s0168-8278(97)80291-7. [DOI] [PubMed] [Google Scholar]
  • 119.Goulis J., Chau T.N., Jordan S., Mehta A.B., Watkinson A., Rolles K. Thrombopoietin concentrations are low in patients with cirrhosis and thrombocytopenia and are restored after orthotopic liver transplantation. Gut. 1999;44:754–758. doi: 10.1136/gut.44.5.754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Panzer S., Seel E., Brunner M., Kormoczi G.F., Schmid M., Ferenci P. Platelet autoantibodies are common in hepatitis C infection, irrespective of the presence of thrombocytopenia. Eur J Haematol. 2006;77:513–517. doi: 10.1111/j.0902-4441.2006.t01-1-ejh2888.x. [DOI] [PubMed] [Google Scholar]
  • 121.Hamaia S., Li C., Allain J.P. The dynamics of hepatitis C virus binding to platelets and 2 mononuclear cell lines. Blood. 2001;98:2293–2300. doi: 10.1182/blood.v98.8.2293. [DOI] [PubMed] [Google Scholar]
  • 122.Kajihara M., Kato S., Okazaki Y., Kawakami Y., Ishii H., Ikeda Y. A role of autoantibody-mediated platelet destruction in thrombocytopenia in patients with cirrhosis. Hepatology. 2003;37:1267–1276. doi: 10.1053/jhep.2003.50209. [DOI] [PubMed] [Google Scholar]
  • 123.Doi T., Homma H., Mezawa S., Kato J., Kogawa K., Sakamaki S., Niitsu Y. Mechanisms for increment of platelet associated IgG and platelet surface IgG and their implications in immune thrombocytopenia associated with chronic viral liver disease. Hepatol Res. 2002;24:23. doi: 10.1016/s1386-6346(02)00010-4. [DOI] [PubMed] [Google Scholar]
  • Zhang W, Nardi MA, Borkowsky W, Karpatkin S. Role of molecular mimicry of hepatitis C virus (HCV) protein with platelet GPIIIa in hepatitis-C related immunologic thrombocytopenia. Blood. In press. [DOI] [PMC free article] [PubMed]
  • 124.Bordin G., Ballare M., Zigrossi P., Bertoncelli M.C., Paccagnino L., Baroli A. A laboratory and thrombokinetic study of HCV-associated thrombocytopenia: a direct role of HCV in bone marrow exhaustion? Clin Exp Rheumatol. 1995;13(Suppl 13):S39–S43. [PubMed] [Google Scholar]
  • 125.de Almeida A.J., Campos-de-Magalhaes M., de Melo Marcal O.P. Hepatitis C virus-associated thrombocytopenia: a controlled prospective, virological study. Ann Hematol. 2004;83:434–440. doi: 10.1007/s00277-004-0844-0. [DOI] [PubMed] [Google Scholar]
  • 126.Iga D., Tomimatsu M., Endo H., Ohkawa S., Yamada O. Improvement of thrombocytopenia with disappearance of HCV RNA in patients treated by interferon-alpha therapy: possible etiology of HCV-associated immune thrombocytopenia. Eur J Haematol. 2005;75:417–423. doi: 10.1111/j.1600-0609.2005.00524.x. [DOI] [PubMed] [Google Scholar]
  • 127.Peck-Radosavljevic M. Thrombocytopenia in liver disease. Can J Gastroenterol. 2000;14(Suppl D) doi: 10.1155/2000/617428. :60D-6D. [DOI] [PubMed] [Google Scholar]
  • 128.McHutchison J.G., Dusheiko G., Shiffman M.L., Rodriguez-Torres M., Sigal S., Bourliere M. Eltrombopag for thrombocytopenia in patients with cirrhosis associated with hepatitis C. N Engl J Med. 2007;357:2227–2236. doi: 10.1056/NEJMoa073255. [DOI] [PubMed] [Google Scholar]
  • 129.Michel M., Cooper N., Jean C., Frissora C., Bussel J.B. Does Helicobater pylori initiate or perpetuate immune thrombocytopenic purpura? Blood. 2004;103:890–896. doi: 10.1182/blood-2003-03-0900. [DOI] [PubMed] [Google Scholar]
  • 130.Gasbarrini A., Franceschi F., Tartaglione R., Landolfi R., Pola P., Gasbarrini G. Regression of autoimmune thrombocytopenia after eradication of Helicobacter pylori. Lancet. 1998;352:878. doi: 10.1016/S0140-6736(05)60004-9. [DOI] [PubMed] [Google Scholar]
  • 131.Maeda S., Ogura K., Yoshida H., Kanai F., Ikenoue T., Kato N. Major virulence factors, VacA and CagA, are commonly positive in Helicobacter pylori isolates in Japan. Gut. 1998;42:338–343. doi: 10.1136/gut.42.3.338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Maeda S., Yoshida H., Ikenoue T., Ogura K., Kanai F., Kato N. Structure of cag pathogenicity island in Japanese Helicobacter pylori isolates. Gut. 1999;44:336–341. doi: 10.1136/gut.44.3.336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Emilia G., Luppi M., Zucchini P., Morselli M., Potenza L., Forghieri F. Helicobacter pylori infection and chronic immune thrombocytopenic purpura: long-term results of bacterium eradication and association with bacterium virulence profiles. Blood. 2007;110:3833–3841. doi: 10.1182/blood-2006-12-063222. [DOI] [PubMed] [Google Scholar]
  • 134.Franceschi F., Christodoulides N., Kroll M.H., Genta R.M. Helicobacter pylori and idiopathic thrombocytopenic purpura. Ann Intern Med. 2004;140:766–767. doi: 10.7326/0003-4819-140-9-200405040-00028. [DOI] [PubMed] [Google Scholar]
  • 135.Takahashi T., Yujiri T., Shinohara K., Inoue Y., Sato Y., Fujii Y. Molecular mimicry by Helicobacter pylori CagA protein may be involved in the pathogenesis of H. pylori-associated chronic idiopathic thrombocytopenic purpura. Br J Haematol. 2004;124:91–96. doi: 10.1046/j.1365-2141.2003.04735.x. [DOI] [PubMed] [Google Scholar]
  • 136.Gerhard M., Rad R., Prinz C., Naumann M. Pathogenesis of Helicobacter pylori infection. Helicobacter. 2002;7(Suppl 1):17–23. doi: 10.1046/j.1523-5378.7.s1.3.x. [DOI] [PubMed] [Google Scholar]
  • 137.Byrne M.F., Kerrigan S.W., Corcoran P.A., Atherton J.C., Murray F.E., Fitzgerald D.J. Helicobacter pylori binds von Willebrand factor and interacts with GPIb to induce platelet aggregation. Gastroenterology. 2003;124:1846–1854. doi: 10.1016/s0016-5085(03)00397-4. [DOI] [PubMed] [Google Scholar]
  • 138.Cines D.B. Inside Blood; ITP: time to “bug off”? Blood. 2007;110:3818–3819. [Google Scholar]
  • 139.Asahi A., Nishimoto T., Okazaki Y., Suzuki H., Masaoka T., Kawakami Y. Helicobacter pylori eradication shifts monocyte Fcgamma receptor balance toward inhibitory FcgammaRIIB in immune thrombocytopenic purpura patients. J Clin Invest. 2008;118:2939–2949. doi: 10.1172/JCI34496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Veneri D., De Matteis G., Solero P., Federici F., Zanuso C., Guizzardi E. Analysis of B- and T-cell clonality and HLA class II alleles in patients with idiopathic thrombocytopenic purpura: correlation with Helicobacter pylori infection and response to eradication treatment. Platelets. 2005;16:307–311. doi: 10.1080/09537100400028685. [DOI] [PubMed] [Google Scholar]
  • 141.Stasi R., Rossi Z., Stipa E., Amadori S., Newland A.C., Provan D. Helicobacter pylori eradication in the management of patients with idiopathic thrombocytopenic purpura. Am J Med. 2005;118:414–419. doi: 10.1016/j.amjmed.2004.09.014. [DOI] [PubMed] [Google Scholar]
  • 142.Miller E., Waight P., Farrington C.P., Andrews N., Stowe J., Taylor B. Idiopathic thrombocytopenic purpura and MMR vaccine. Arch Dis Child. 2001;84:227–229. doi: 10.1136/adc.84.3.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Abramson J.S., Pickering L.K. US immunization policy. JAMA. 2002;287:505–509. doi: 10.1001/jama.287.4.505. [DOI] [PubMed] [Google Scholar]
  • 144.Black C., Kaye J.A., Jick H. MMR vaccine and idiopathic thrombocytopaenic purpura. Br J Clin Pharmacol. 2003;55:107–111. doi: 10.1046/j.1365-2125.2003.01790.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Schattner A. Consequence or coincidence?: The occurrence, pathogenesis and significance of autoimmune manifestations after viral vaccines. Vaccine. 2005;23:3876–3886. doi: 10.1016/j.vaccine.2005.03.005. [DOI] [PubMed] [Google Scholar]
  • 146.Neau D., Bonnet F., Michaud M., Perel Y., Longy-Boursier M., Ragnaud J.M. Immune thrombocytopenic purpura after recombinant hepatitis B vaccine: retrospective study of seven cases. Scand J Infect Dis. 1998;30:115–118. doi: 10.1080/003655498750003465. [DOI] [PubMed] [Google Scholar]
  • 147.Naidu S., Messmore H., Caserta V., Fine M. CNS lesions in neonatal isoimmune thrombocytopenia. Arch Neurol. 1983;40:552–554. doi: 10.1001/archneur.1983.04050080052009. [DOI] [PubMed] [Google Scholar]
  • 148.Beeler J., Varricchio F., Wise R. Thrombocytopenia after immunization with measles vaccines: review of the vaccine adverse events reporting system (1990 to 1994) Pediatr Infect Dis J. 1996;15:88–90. doi: 10.1097/00006454-199601000-00020. [DOI] [PubMed] [Google Scholar]
  • 149.Nieminen U., Peltola H., Syrjala M.T., Makipernaa A., Kekomaki R. Acute thrombocytopenic purpura following measles, mumps and rubella vaccination: A report on 23 patients. Acta Paediatr. 1993;82:267–270. doi: 10.1111/j.1651-2227.1993.tb12657.x. [DOI] [PubMed] [Google Scholar]
  • 150.Jadavji T., Scheifele D., Halperin S. Thrombocytopenia after immunization of Canadian children, 1992 to 2001. Pediatr Infect Dis J. 2003;22:119–122. doi: 10.1097/01.inf.0000048961.08486.d1. [DOI] [PubMed] [Google Scholar]
  • 151.Chou A.L., Huang W.W., Tsao S.M., Li C.T., Su C.C. Human herpesvirus type 8 in patients with cirrhosis: correlation with sex, alcoholism, hepatitis B virus, disease severity, and thrombocytopenia. Am J Clin Pathol. 2008;130:231–237. doi: 10.1309/4FMBN316N4792UQ4. [DOI] [PubMed] [Google Scholar]
  • 152.Hui D.S. Review of clinical symptoms and spectrum in humans with influenza A/H5N1 infection. Respirology. 2008;13(Suppl 1):S10–S13. doi: 10.1111/j.1440-1843.2008.01247.x. [DOI] [PubMed] [Google Scholar]
  • 153.Rafailidis P.I., Mourtzoukou E.G., Varbobitis I.C., Falagas M.E. Severe cytomegalovirus infection in apparently immunocompetent patients: a systematic review. Virol J. 2008;5:47. doi: 10.1186/1743-422X-5-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Yang M., Ng M.H., Li C.K. Thrombocytopenia in patients with severe acute respiratory syndrome (review) Hematology. 2005;10:101–105. doi: 10.1080/10245330400026170. [DOI] [PubMed] [Google Scholar]
  • 155.Vipan W. Quinine as a cause of purpura. Lancet. 1865;2:37. [Google Scholar]
  • 156.Drug-induced thrombocytopenia. http://moon.ouhsc.edu/jgeorge/DITP.html Accessed September 11, 2008.
  • 157.Aster R.H., Bougie D.W. Drug-induced immune thrombocytopenia. N Engl J Med. 2007;357:580–587. doi: 10.1056/NEJMra066469. [DOI] [PubMed] [Google Scholar]
  • 158.van den Bemt P.M., Meyboom R.H., Egberts A.C. Drug-induced immune thrombocytopenia. Drug Saf. 2004;27:1243–1252. doi: 10.2165/00002018-200427150-00007. [DOI] [PubMed] [Google Scholar]
  • 159.Visentin G.P., Newman P.J., Aster R.H. Characteristics of quinine- and quinidine-induced antibodies specific for platelet glycoproteins IIb and IIIa. Blood. 1991;77:2668–2676. [PubMed] [Google Scholar]
  • 160.Asvadi P., Ahmadi Z., Chong B.H. Drug-induced thrombocytopenia: localization of the binding site of GPIX-specific quinine-dependent antibodies. Blood. 2003;102:1670–1677. doi: 10.1182/blood-2002-07-2175. [DOI] [PubMed] [Google Scholar]
  • 161.Parker C.W. Hapten immunology and allergic reactions in humans. Arthritis Rheum. 1981;24:1024–1036. doi: 10.1002/art.1780240808. [DOI] [PubMed] [Google Scholar]
  • 162.Bougie D.W., Wilker P.R., Aster R.H. Patients with quinine-induced immune thrombocytopenia have both “drug-dependent” and “drug-specific” antibodies. Blood. 2006;108:922–927. doi: 10.1182/blood-2006-01-009803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Aster R.H. Immune thrombocytopenia caused by glycoprotein IIb/IIIa inhibitors. Chest. 2005;127:53S–59S. doi: 10.1378/chest.127.2_suppl.53S. [DOI] [PubMed] [Google Scholar]
  • 164.McCorry R.B., Johnston P. Fatal delayed thrombocytopenia following abciximab therapy. J Invasive Cardiol. 2006;18:E173–E174. [PubMed] [Google Scholar]
  • 165.Curtis B.R., Swyers J., Divgi A., McFarland J.G., Aster R.H. Thrombocytopenia after second exposure to abciximab is caused by antibodies that recognize abciximab-coated platelets. Blood. 2002;99:2054–2059. doi: 10.1182/blood.v99.6.2054. [DOI] [PubMed] [Google Scholar]
  • 166.Tcheng J.E., Kereiakes D.J., Lincoff A.M., George B.S., Kleiman N.S., Sane D.C. Abciximab readministration: results of the ReoPro Readministration Registry. Circulation. 2001;104:870–875. doi: 10.1161/hc3301.094533. [DOI] [PubMed] [Google Scholar]
  • 167.Curtis B.R., Divgi A., Garritty M., Aster R.H. Delayed thrombocytopenia after treatment with abciximab: a distinct clinical entity associated with the immune response to the drug. J Thromb Haemost. 2004;2:985–992. doi: 10.1111/j.1538-7836.2004.00744.x. [DOI] [PubMed] [Google Scholar]
  • 168.von dem Borne A.E., Pegels J.G., van der Stadt R.J., van der Plas-van Dalen C.M., Helmerhorst F.M. Thrombocytopenia associated with gold therapy: a drug-induced autoimmune disease? Br J Haematol. 1986;63:509–516. doi: 10.1111/j.1365-2141.1986.tb07528.x. [DOI] [PubMed] [Google Scholar]
  • 169.Garner S.F., Campbell K., Metcalfe P., Keidan J., Huiskes E., Dong J.F. Glycoprotein V: the predominant target antigen in gold-induced autoimmune thrombocytopenia. Blood. 2002;100:344–346. doi: 10.1182/blood.v100.1.344. [DOI] [PubMed] [Google Scholar]
  • 170.George J.N., Raskob G.E., Shah S.R., Rizvi M.A., Hamilton S.A., Osborne S. Drug-induced thrombocytopenia: a systematic review of published case reports. Ann Intern Med. 1998;129:886–890. doi: 10.7326/0003-4819-129-11_part_1-199812010-00009. [DOI] [PubMed] [Google Scholar]
  • 171.Hirsh J., Heddle N., Kelton J.G. Treatment of heparin-induced thrombocytopenia: a critical review. Arch Intern Med. 2004;164:361–369. doi: 10.1001/archinte.164.4.361. [DOI] [PubMed] [Google Scholar]
  • 172.Warkentin T.E., Kelton J.G. A 14-year study of heparin-induced thrombocytopenia. Am J Med. 1996;101:502–507. doi: 10.1016/s0002-9343(96)00258-6. [DOI] [PubMed] [Google Scholar]
  • 173.Lubenow N., Kempf R., Eichner A., Eichler P., Carlsson L.E., Greinacher A. Heparin-induced thrombocytopenia: temporal pattern of thrombocytopenia in relation to initial use or reexposure to heparin. Chest. 2002;122:37–42. doi: 10.1378/chest.122.1.37. [DOI] [PubMed] [Google Scholar]
  • 174.Warkentin T.E., Kelton J.G. Temporal aspects of heparin-induced thrombocytopenia. N Engl J Med. 2001;344:1286–1292. doi: 10.1056/NEJM200104263441704. [DOI] [PubMed] [Google Scholar]
  • 175.Warkentin T.E., Kelton J.G. Delayed-onset heparin-induced thrombocytopenia and thrombosis. Ann Intern Med. 2001;135:502–506. doi: 10.7326/0003-4819-135-7-200110020-00009. [DOI] [PubMed] [Google Scholar]
  • 176.Reilly M.P., Taylor S.M., Hartman N.K., Arepally G.M., Sachais B.S., Cines D.B. Heparin-induced thrombocytopenia/thrombosis in a transgenic mouse model requires human platelet factor 4 and platelet activation through FcgammaRIIA. Blood. 2001;98:2442–2447. doi: 10.1182/blood.v98.8.2442. [DOI] [PubMed] [Google Scholar]
  • 177.Rauova L., Poncz M., McKenzie S.E., Reilly M.P., Arepally G., Weisel J.W. Ultralarge complexes of PF4 and heparin are central to the pathogenesis of heparin-induced thrombocytopenia. Blood. 2005;105:131–138. doi: 10.1182/blood-2004-04-1544. [DOI] [PubMed] [Google Scholar]
  • 178.Rauova L., Zhai L., Kowalska M.A., Arepally G.M., Cines D.B., Poncz M. Role of platelet surface PF4 antigenic complexes in heparin-induced thrombocytopenia pathogenesis: diagnostic and therapeutic implications. Blood. 2006;107:2346–2353. doi: 10.1182/blood-2005-08-3122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Cines D.B., Tomaski A., Tannenbaum S. Immune endothelial-cell injury in heparin-associated thrombocytopenia. N Engl J Med. 1987;316:581–589. doi: 10.1056/NEJM198703053161004. [DOI] [PubMed] [Google Scholar]
  • 180.Arepally G.M., Mayer I.M. Antibodies from patients with heparin-induced thrombocytopenia stimulate monocytic cells to express tissue factor and secrete interleukin-8. Blood. 2001;98:1252–1254. doi: 10.1182/blood.v98.4.1252. [DOI] [PubMed] [Google Scholar]
  • 181.Thachil J. Heparin induced thrombocytopenia with thrombosis: a two step process? Hematology. 2008;13:181–182. doi: 10.1179/102453308X316176. [DOI] [PubMed] [Google Scholar]
  • 182.Bauer T.L., Arepally G., Konkle B.A., Mestichelli B., Shapiro S.S., Cines D.B. Prevalence of heparin-associated antibodies without thrombosis in patients undergoing cardiopulmonary bypass surgery. Circulation. 1997;95:1242–1246. doi: 10.1161/01.cir.95.5.1242. [DOI] [PubMed] [Google Scholar]
  • 183.Warkentin T.E., Sheppard J.I., Moore J.C., Sigouin C.S., Kelton J.G. Quantitative interpretation of optical density measurements using PF4-dependent enzyme-immunoassays. J Thromb Haemost. 2008;6:1304–1312. doi: 10.1111/j.1538-7836.2008.03025.x. [DOI] [PubMed] [Google Scholar]
  • 184.Greinacher A., Eichler P., Lubenow N., Kiefel V. Drug-induced and drug-dependent immune thrombocytopenias. Rev Clin Exp Hematol. 2001;5:166–200. doi: 10.1046/j.1468-0734.2001.00041.x. [DOI] [PubMed] [Google Scholar]
  • 185.Liebman H. Other immune thrombocytopenias. Semin Hematol. 2007;44:S24–S34. doi: 10.1053/j.seminhematol.2007.11.004. [DOI] [PubMed] [Google Scholar]

Articles from Seminars in Hematology are provided here courtesy of Elsevier

RESOURCES